You are on page 1of 40

Accepted Manuscript

A systematic reconsideration on proteases

Poorani Gurumallesh, Kamalini Alagu, Baskar Ramakrishnan,


Shanmugaprakash Muthusamy

PII: S0141-8130(18)35199-7
DOI: https://doi.org/10.1016/j.ijbiomac.2019.01.081
Reference: BIOMAC 11514
To appear in: International Journal of Biological Macromolecules
Received date: 3 October 2018
Revised date: 3 January 2019
Accepted date: 18 January 2019

Please cite this article as: Poorani Gurumallesh, Kamalini Alagu, Baskar Ramakrishnan,
Shanmugaprakash Muthusamy , A systematic reconsideration on proteases. Biomac
(2018), https://doi.org/10.1016/j.ijbiomac.2019.01.081

This is a PDF file of an unedited manuscript that has been accepted for publication. As
a service to our customers we are providing this early version of the manuscript. The
manuscript will undergo copyediting, typesetting, and review of the resulting proof before
it is published in its final form. Please note that during the production process errors may
be discovered which could affect the content, and all legal disclaimers that apply to the
journal pertain.
ACCEPTED MANUSCRIPT

A systematic reconsideration on proteases

Poorani Gurumallesh, Kamalini Alagu, Baskar Ramakrishnan*, Shanmugaprakash Muthusamy

Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, India

PT
Corresponding author

RI
Dr. R. Baskar

Associate Professor

SC
Kumaraguru College of Technology NU
Chinnavedampatti, Saravanampatty

Coimbatore – 641049
MA

Email: baskar.r.bt@kct.ac.in

Phone: +91 9894709273


D

+91 422 2661100


E
PT
CE
AC

Conflict of Interest: The authors declare no conflict of interest


ACCEPTED MANUSCRIPT

Abstract

Proteases are a group of large complex enzyme molecules that perform highly focused proteolysis
functions. A vast quantity of the protease enzymes is predominantly sourced from microbial
fermentation process, although proteases tend to natively present in plant, animals and humans.
Proteases possess a pervasive importance in medical and pharmaceutical sector, because of its enriched
specificity towards biomolecules. They are also actively encompassed in regulating certain
physiological pathways. A distinct territory of human disorders is treated by substrate specific
proteases. Enormous numbers of catalytic activities in habitual metabolism process of a living organism

PT
are protease dependent. Pilot scale researches and product development in industrial biotechnology
sectors are wholly based on any one of the protease enzymes. The applications of the protease enzymes

RI
and its economic benefits of being an eco-friendly material are far-reaching. This review presents a
brief overview on the classification and sources of various types of proteases. We describe the essential

SC
evidences of role of protease in different sectors. The proteases could be a potential relieves to harmful
synthetic chemicals in distinctive industrial processes and thus gains global perception.
NU
Keywords: Proteases; Categorization; Industrial application.
MA
E D
PT
CE
AC
ACCEPTED MANUSCRIPT

1. Overview

The peptide bonds in between amino acids and proteins are hydrolysed by proteases. Around 60%
enzymes in the global market accounts to proteases [1]. The catalytic activity of the protease are
involved in various processes like digestion, apoptosis and so on. Proteases are so much indispensable
in all living organisms for synthesis of necessary biomolecules and to control the size, shape, turnover
and the composition of fundamental proteins[2]. The International protease network states that every
human hold closely 500 proteases accounting to 2% of total genome. However, the metabolic role of
certain proteases in the human system are yet to be studied [3]. The highly specific and limited cleavage

PT
in the substrate by the protease is one among the foremost metabolic reactions in all of the living
organisms. Hydrolysis by protease is an originator to various biological process such as DNA

RI
replication and cell cycle [4].

SC
Proteases are classified by its diverse attributes. Proteases were acquired majorly from various plant
and animal sources before the exploitation of the microbes. In a while, the plants and animals confronted
hitches in its continued existence due to climatic factors and cumulative devastation of natural habitats
NU
by humans. The ethical issues were on rise upon sacrificing animals to isolate protease. Thereby the
microbes arrive at attention for researchers to produce large scale protease enzyme at high yield rates.
MA

Almost two-third of the commercial proteases available in the market all over the world belongs to
proteases from microorganisms [5].

Proteases are one of the most experimental enzymes and plays decisive role at different sectors such as
D

commercial, pharmaceutical, analytical, diagnostic and effluent abatement. In this review, the
E

comprehensive description of classification of protease, sources of protease enzyme and the potential
PT

utilizations of the enzyme in a variety of industries will be discussed. The overview of the review article
is illustrated in the graphical abstract (Figure 1).
CE

2. Categorization of Proteases
AC

The IUBMB Nomenclature Committee states that classification of proteases could be made under
subgroup 4 of group 3 (hydrolases) [6]. The grouping of proteases is based on various strategies such
as the reaction type where protease acts as a catalyst; the nature and the chemical properties of the
catalytic site and the evolution of the protease structure and their relationship [7]. The organisation of
protease based on their specificity or catalytic mechanism was discussed by Agarwal [8]. Protease (E.C
No.3.4) enzyme that hydrolyse the peptide bonds are further sectioned into Exopeptidases (E.C. No
(3.4.11 -3.4.19) and Endopeptidases (E.C. No (3.4.21-3.4.24, 3.4.99) depending upon the type of the
reaction [9].

2.1 Exopeptidases
ACCEPTED MANUSCRIPT

Exopeptidases executes its enzyme activity by hydrolysis merely on the nitrogen or carbon terminal
points of the substrate chain entirely made up of polypeptide [10]. Exopeptidases are further sorted
conferring to the specificity such as the size of the fragment, identity of the liberated fragment, terminus
of the origin and the size restriction on the length of the specific peptide chain [8]. Exopeptidases acts
in two forms, namely Aminopeptidase and Carboxypeptidase.

As the name indicates, the aminopeptidases put on its activity at a free N terminal point of the protein
and as a result of digestion it releases a mono amino acid residue, a dipeptide, or a tripeptide [11]. The

PT
presence of aminopeptidases is reported in a wide variation of microbial species that includes bacteria
as well as fungi, plants and animals. The aminopeptidases are secreted as intracellular and extracellular

RI
aminopeptidases [12]. Methionine aminopeptidases (MAP) accomplishes crucial cellular functions,
which shares homology with a vast range organism, and functions by cleaving N-terminal methionine

SC
from freshly synthesized proteins [13].
NU
The C terminals of the protein chain is majorly influenced by carboxypeptidases. Depending on the
character of the residual amino acid at the active site, carboxypeptidases are clustered under three
classifications: serine carboxypeptidases, cysteine carboxypeptidases, and metallo carboxypeptidases
MA

[14]. The first generation carboxypeptidases such as pancreatic carboxypeptidase A1 and A2, have their
potentiality exhibited at the process of digestion of food molecules. The other properties revealed by
the carboxypeptidases are their active participation in healing of internal and external wound and blood
D

clotting. The classification of proteases is outlined in Table 1.


E

Table 1: Categorization of proteases


PT

TYPE OF EC.NO MECHANISM EXAMPLES


PROTEASE
Aminopeptidases 3.4. Acts on free N-terminus Cleaves one amino Peptidase_M
CE

of a protein acid and N-terminal 1


Dipeptidases 3.4.13 Hydrolyses the Cleaves two amino Aspartyl
AC

dipeptide bond at the N- acid and N-terminal dipeptidase


terminus
Tripeptidases 3.4.11 Hydrolyses the Cleaves three amino Lympho
tripeptide bond at the N- acid and N-terminal peptidase
terminus
Carboxypeptidases 3.4.16- Acts on free C-terminus Cleaves one amino Pancreatic
3.4.18 and releases a single acid from C-terminal carboxypepti
amino acid dase A
ACCEPTED MANUSCRIPT

Serine type protease 3.4.16 Serine serves as a Cleaves peptide Subtilisin


nucleophilic amino acid bonds with its partial
at active site negative oxygen
atom
Metallo protease 3.4.17 Catalytic mechanism Cleaves the C- Thermolysin
involves a metal, terminus, the
often zinc unbound COOH
group

PT
Cysteine type 3.4.18 Catalytic mechanism Deprotonation of a Papain
protease involves a nucleophilic thiol followed by

RI
cysteine thiol nucleophilic attack

SC
2.2 Endopeptidases
Endopeptidases are proactively involved in causing cleavage in amino acids that are non-terminal [15].
NU
The endopeptidases are subjected to classification reliant on the chemical kind of the group that is
primarily in charge for the catalytic activity. There are six classes of protease; (i) Cysteine proteases,
(ii) Serine proteases, (iii) Threonine proteases, (iv) Glutamic acid proteases, (v) Aspartic acid proteases
MA

and (vi) Metalloproteases.

Serine protease are identified by the occurrence of residual serine in the catalytic binding site [16] .
D

Serine proteases are sorted into 20 families depending upon their resemblances in their structures.
E

Devising a common ancestor, the families are additionally segmented into six class [11]. Every single
PT

serine protease encloses three residual molecules at their catalytic binding site: aspartate, histidine and
serine. Serine proteases contribute to one third of all known proteases [17]. The mechanism of serine
peptidase is as simple as fragment transfer like other group of enzymes, although in this case the transfer
CE

is to water [18]. Considering a deep look into the mechanism of serine protease, almost all of them
retain a basic dyad concept in which Lys or His found to be bound with the catalytic Ser. Other serine
AC

proteases provoke catalysis through novel triads of residues, such as a pair of His residues combined
with the nucleophilic Ser [17].

Aspartic acid protease are the acidic proteases that use an activated water molecule constrained to
aspartate residues at the catalytic site of action [19]. Those proteases are settled into three families
namely retropepsin, pararetroviruses proteins and pepsin. As per the noted activity of aspartic acid
proteases, the functional group of the enzyme would be targeted. While targeting, there is no nucleopilic
attack happening around and also there is absence of formation of covalent substance that was expected
to occur in between the enzyme and a substrate fragment [18]. The examples of potential aspartic acid
ACCEPTED MANUSCRIPT

proteases include pepsin, cathepsin and rennin. Aspartic acid proteases are highly active and efficient
at minimal pH range (3 - 4).

Cysteine proteases provide the validated targets to treat a wide range of human disorders. Prokaryotes
and eukaryotes contain proteases that have cysteine at their active site [20]. The catalytic dyad of
cysteine and histidine are the active components that are crucial for cysteine proteases to exhibit its
potentiality. About 20 families of cysteine proteases have been recognized [14]. Cysteine proteases are
broadly divided into four groups based on side chain specificity: (i) papain-like, (ii) trypsin-like with

PT
preference for cleavage at the arginine residue, (iii) specific to glutamic acid, and (iv) others (20).
Calpains are a type of cysteine proteases which need calcium ions for enzyme activation. 15 isoforms

RI
of the calpain were reported and 11 of them have been identified in humans. µ-Calpain and m-calpain
are the two types of calpain that have been isolated which differ in their calcium requirement [11].

SC
Cysteine proteases are active only in the presence of reducing agents such as HCN or cysteine.
NU
Metalloproteases belong to the diverse group of the catalytic types of proteases. Every single
metalloprotease requires a divalent metal ion to exhibit its proteolytic activity on the specific protease.
There are about 30 families identified so far [21]. Metalloproteases can be divided into four groups
MA

depending upon the specificity of their action, (i) neutral, (ii) alkaline, (iii) Myxobacter I, and (iv)
Myxobacter II. In one way, metalloprotease is comparable to aspartic peptidases so that they do not
form the covalent intermediate, hence they need a metal ion to exert this effect.
E D

Proteolytic enzyme with a threonine residue in its active side are said to be threonine proteases. To
PT

perform catalysis, the threonine proteases use a nucleophile which is primarily a secondary alcohol of
the N-terminal threonine. Enzyme catalysis by threonine protease involves two stages. Initially, the
substrate is targeted by the nucleophile to form acyl-enzyme intermediate, followed by water dependent
CE

hydrolysis to release the products and free enzymes [22].


AC

Glutamic proteases are characterized in such a way that; all active site contains a glutamic acid residue.
The first ever elucidated structure of the enzyme was scytalidoglutamic protease, where the active
contained a catalytic dyad (Glutamic acid and Glutamine). Glutamic proteases are acidic in nature and
works better at pH 2. Eqolisin is a potential example of glutamic protease which was isolated from the
fungi Scytalidium lignicola [23]. The common characters shared by the different endoproteases are
given in Table 2.

Table 2: Common characteristics of endopeptidases


ACCEPTED MANUSCRIPT

Type of protease Serine protease Cysteine / Thiol Aspartic/ Metallo protease


Trypsin protease Carboxyl
Chymotrypsin µ-Calpain protease
m- Calpain Pepsin
Cathapsin B H L Cathapsin D
EC No 3.4.21 3.4.22 3.4.23 3.4.24
Active site of Serine, Histidine Aspartate/ Aspartate/ Phenyl-alanine/
amino acid and Dispartate Cysteine Cysteine Leucine
2+ 2+
Metal Ions Ca Ca Zn2+, Ca2+

PT
Optimum pH 6-11 2-3 2-5 5-8
Temperature 50-70 40-55 40-55 65-85

RI
(ºC)
Molecular weight 18-35 20-37 30-48 19-37

SC
(KDa) 69-74
Mechanism Endopeptidase Endopeptidase Endopeptidase Endopeptidase
NU
have an active have an active have an active have an active
center, serine center, cysteine center, Aspartic center, metal ions
involved in involved in involved in (Zn+) involved in
MA

catalytic process catalytic process catalytic process catalytic process


Application Blood clotting, Apoptosis of Blood pressure Tumor
immunity and breast cancer cells control; metastasis;
D

inflammation Digestive Wound healing;


E

response; enzyme; Meat Embryonic


PT

Digestive tenderizer development


enzymes
CE

3. Sources of Proteases
AC

The occurrence of protease is reported in all organisms, like prokaryotes, eukaryotes, plants and
animals. The type of protease secreted is completely dependent on the host and its adaptability to its
immediate environment. A large number of factors must be taken into consideration for fermentative
production of protease from the microbial source. The detailed description of different source of
proteases is given below with examples.

3.1 Plants
The plants as a source of proteases has a lot many considerations that include dependency on suitable
climatic conditions and availability of agricultural land for cultivation. Plants respond to the
environment in such a manner that the environmental signals play a crucial role in normal development
ACCEPTED MANUSCRIPT

pathways. The time utilization and labour involvement are the factors to be pondered before electing a
plant as a source to extract protease [24]. In plants, proteases are partaking in the events like
mobilization of storage proteins, apoptosis, etc. Proteases are generally inactive proenzyme fragments
at N-terminal ends of a precursor [25]. Predominantly, plant proteases are cysteine endopeptidases and
rarely other proteases are observed [26]. Bromelain, papain, ficin and keratinases are a few of the
illustrious proteases of plant origin.
Miliin, is a serine protease extracted from latex of Euphorbia milii. The protease exhibited maximal
activity around pH 9 and at 35ºC. The molecular weight determined was about 75 kDa. Miliin is broadly

PT
used as a molluscicidal agent and as antithrombic drugs [27]. The another type of miliin retrieved from
the Euphorbia species is a thiol dependant serine protease. The 79 kDa enzyme demonstrated activity

RI
at 60ºC and the active pH range is 7.5-11 [28].

SC
Carnivorous plants also secrete protease that of huge biotechnological and pharmaceutical importance.
Drosera capensis is one such plant that has a large number of genes that codes for cysteine proteases.
NU
Upon sequence comparison, cysteine proteases from Drosera species showed homology to various
plant proteases [29]. Wrightin, a known serine protease is obtained from the Wrightia tinctoria latex.
The enzyme having a molecular weight of 57.9 kDa is active at pH 7.5-10 and temperature of 70ºC.
MA

Wrightin is a potential candidate for food industries [30].

Papain is a plant cysteine protease isolated from Carica papaya latex fruits. The papain has broader
D

specificity towards substrates because of presence of numerous isoenzymes within it. Papain is active
E

between pH 5 – 9 and found to be stable till 80ºC. The papain plays a key role in varied biological
PT

process and used comprehensively in meat tenderizing industry, drug designing and so on [31]. Carnein
is a serine protease from the latex of Ipomea carnea. The molecular mass is 80.24 kDa and optimal
conditions for high protease activity are pH 6.5 and 65ºC [32]. An aspartic proteases exhibiting different
CE

physiological roles is known to be purified from the potato leaves [33]. A thiol dependant protease is
reported in crown leaf of pineapple [34] and a neutral protease from the leaves of Raphanus sativus is
AC

purified and used in various industrial applications [35].

Each plants express protease under different environmental conditions. In parsley leaves, a 70 kDa
serine protease is observed, where the protease activity is found to increase with advancement in
senescence [36]. Similarly, protease from barley and alfalfa establishes degradation of protein only
during the senescence of the foliar region [37]. Cathepsin B, a cysteine protease from a Himalayan plant
Picrorhiza kurrooa is a 39 kDa protein. The protease is expressed with response to phytohormones that
upregulate the leaf senescence [38]. A group of cysteine proteases C1A are secreted in abundance only
during the leaf senescence. This shows how the plants are dependent on the environment and physiology
to express certain proteins [39].
ACCEPTED MANUSCRIPT

Bromelain is an industrially important cysteine protease enzyme obtained from the pineapple parts such
as stem and juices. The active range of enzyme is from pH 5 to 9 and optimum temperature is 70°C.
The optimum temperature is found to be lesser than that of papain. A review by Barun on bromelain
demonstrates the potential applications of the cysteine protease in wide range of industries [40]. The
complete information of plant based protease, source, purification strategies and applications are
described in a review article by Cecilia and Xavier [18].

PT
3.2 Animals

Very few economically important proteases are originated from animal species. Rennin, chymotrypsin,

RI
pancreatic trypsin, pepsins are the animal proteases that play a fundamental role in digestive process.
Animal proteases are isolated in bulk quantities depending on the existence of livestock slaughter. The

SC
extraction is however governed by various policies categorized under political and agricultural sectors
[24]. In order to look for an alternative to minimise the animal slaughter, recombinant DNA technology
NU
would be performed for massive production of animal proteases. Also, the animal proteases gain
attention to be a bio-control measures for insect pests.
MA

A serine protease, Trypsin is the main intestinal digestive enzyme present widely in vertebrates. Trypsin
is an important biomolecule that could hydrolyse the proteins present in food. Trypsin acts on the
D

carboxyl end of the amino acids lysine or arginine. Trypsin from bovine and porcine sources is of 23.3
E

kDa molecular weight and works best at 37ºC. Trypsin is extensively used in a tissue culture lab for the
process of harvesting the cells. Apart from this, trypsin is used as medicinal and food industries [41].
PT

Chymotrypsin is yet another serine protease found in pancreatic juice of an animal. The mechanism of
CE

action is that, chymotrypsin attacks the unreactive carbonyl with a help of a nucleophile. Chymotrypsin
does not exhibit allosteric effects. For diagnostic and analytical purposes pure chymotrypsin which is
AC

an expensive enzyme is utilized. Chymotrypsin exerts its specificity for hydrolysis of amino acids such
as phenylalanine, tyrosine, or tryptophan [42]. Chymotrypsin is present in precursor form
chymotrypsinogen. Through a cascade of processes, the inactive precursor is converted into
chymotrypsin by trypsin. A large volume de-allergenizing of milk protein is conceded by chymotrypsin.

An acidic aspartyl protease called Pepsin is existing in the stomachs of almost all vertebrates. The
precursor of the pepsin is a zymogen namely pepsinogen. Hydrochloric acid from the parietal cells is
required to activate the zymogen. Pepsin potentially cleaves the amino acids such as phenylalanine,
tryptophan and tyrosine at the N-terminal side. Pepsin has been extracted from Japanese monkey, goat,
rat and rabbit. A review of protease from fish is given in detail by Lisha [43].
ACCEPTED MANUSCRIPT

3.3 Microbes

Microbial proteases contribute to nearly two-third of industrially essential proteases. Only certain
microorganisms manufacture proteases with anticipated qualities. The growth and maintenance of the
host microbes when compared to plant and animals is quite stress-free and less time consuming. Some
microorganism has their natural capability to secrete protease by themselves without any induction.
Proteases are produced in large volume by fermentation process of microorganisms in a planned

PT
bioreactor. Two approaches, solid state and submerged fermentation are exercised to extract protease
from microbes. Among the two processes, solid state fermentation is preferred more, because of low

RI
consumption of energy. The protease and its characters extracted from different microorganisms are
outlined in Table 3.

SC
Table 3: Microbial proteases from bacteria and its characteristics
NU
Micro - Source of Protease Purification Optimal Application Ref.
organism Isolation Type strategy conditions
MA

Bacillus Gut of Chymo Crude protein pH 7.8 Digestion [44]


subtilis Helicoverp trypsin like Temp of
RTSBA 6 a armigera protease 35ºC proteinacio
D

us food
E

Bacillus Agro – De-hairing ASP, Sephadex PH 6-9 Detergent [45]


PT

cereus strain industrial alkaline G-75 column Temp and leather


AT residues protease chromatography 50ºC industry
and cow
CE

dung
Bacillus Saltern Alkaline Crude Protein pH 8 - [46]
AC

licheniformis sediments proteases Temp


37ºC
Bacillus Soil Thermostabl ASP, DEAE – pH 7.5 De-hairing [47]
megatarium e alkaline cellulose ion Temp of skin
proteases exchange 50ºC
chromatography
and Sephadex
G-200 gel
filtration
ACCEPTED MANUSCRIPT

Bacillus Mangroves Alkaline ASP, SDS-Page pH 7 Detergent [48]


circulans proteases Temp additive
BM15 40ºC
Bacillus Pigeon pea Cellulase Crude protein pH 11 Food [49]
species JB-99 waste as free Temp industry
substrate thermostable 70ºC
protease
Pseudomona Tannery Salt tolerant ASP pH 9 Leather [50]

PT
s aeruginosa saline protease Temp industry
strain BC1 waste water 30ºC

RI
Listeria Degraded Alkaline Crude Protein pH 9 Therapeutic [51]
monocytogen meat of Protease Temp application

SC
es cow 30ºC s
Bacillus Soil of a Organic Crude Protein pH 10 Potential [52]
NU
cereus strain wood solvent Temp use in
146 factory tolerant 37ºC industries
alkaline
MA

protease
Halotolerant Coastal Metalloprote ASP and pH 9 Stain [53]
alkaliphilic sediment of ase Dialysis Temp removal
D

Bacillus Goa 37ºC capability


E

cereus CS1
PT

Geobacillus Sewage Thermoactiv Crude Protein pH 8-9 - [54]


caldoproteol sludge e Temp
yticus extracellular 70ºC
CE

protease
Bacillus - Serine Sephadex G-200 pH 9 Blood stain [55]
AC

laterosporus protease gel filtration; Temp removal


DEAE cellulose 75ºC
chromatography
Pseudomona Meat waste Alkaline Crude Protein pH 9 - [56]
s fluorescens contaminat protease Temp
e soil 37ºC

The microbes that are employed in fermentative production of protease are briefly explained in a review
article by Kanupriya et al [57]. Protease producing microorganisms are isolated from various sources
like soil, vegetable wastes, milk processing plant [58], and poultry waste contaminated soil [59] and so
ACCEPTED MANUSCRIPT

on. Every microorganism needs an optimized growth media and environmental conditions to secrete
the protease enzyme. Once secreted, a series of downstream processing steps must be carried out in
order to obtain pure fraction of the protease enzyme. A brief outline of the protease purification steps
is demonstrated in figure 2.

The next group of microbes to be focused for protease production are the fungi. Filamentous fungi have
an extraordinary capability to secrete functionally active proteins. Trichoderma and Aspergillus are two
dominant fungal strains utilized for industrial protease production. Pleurotus citrinopileatus is an edible

PT
mushroom that acts as a rich source of alkaline protease [60]. The other fungal species involved in
protease production are described in Table 4.

RI
Table 4: Microbial proteases from fungi and its characteristics

SC
Microorganism Source of Protease Purification Optimal Applications Ref.
Isolation Type strategy conditions
NU
Rhizopus oryzae - Aspartic Anion pH 3.4 Food [61]
protease exchange Temp processing
chromatogra 75ºC industry
MA

phy
Lentinus citrinus Ligno Neutral Crude pH 7 Food [62]
cellulosic protease protein Temp industry
D

waste 50ºC
E

Rhizomucor Local Rennet Crude Temp Milk [63]


PT

nainitalensis isolates protein 40ºC coagulation


Engyodontium Marine Alkaline ASP ion- pH 11 Detergent [64]
album sediment protease exchange Temp industry
CE

chromatogra 60ºC
phy
AC

Beauveria felina Soy Alkaline Crude pH 9 Hydrolysis of [65]


protein protease protein Temp soy protein
isolate 55ºC
Penicillium Soil Alkaline Crude pH 8 Detergent [66]
godlewskii SBSS protease protein Temp industry
25 35ºC

Apart from the above mentioned fungi species there are lot many fungal sources that secrete
commercially important protease enzyme. That includes Mucor species [67], Aspergillus clavatus [68],
Penicillium chrysogenum [69] and so on. Proteases from certain actinomycetes also comes under
ACCEPTED MANUSCRIPT

microbial proteases. Actinomycetes are a group of bacteria with morphology similar to the fungi.
Streptomyces nogalator is a protease producing actinomycete, isolated from the soil of leather and hair
dumping areas. The enzyme was found to be useful in depilation of goat skin [70]. Another alkali-
thermotolerant protease was isolated from Streptomyces sp DP2, which exhibited maximal activity over
at pH 8 and possessed thermostability at 90ºC [71].

4. Recent advances and strategies to improvise protease production and yield

PT
Periodically, gene knockdown and genetic manipulation have been undertaken through various
technologies. There are certain ineffectiveness and drawbacks on adopting those conventional

RI
methodologies as we do not possess any control over prevailing protein production levels. In order to
achieve strategic enzyme production, synthetic biology tools are to be hired. Among the tools, CRISPR

SC
– Clustered Regularly Interspaced Short Palindromic Repeats, are the most effective with high
potentiality in achieving gene modification [72]. The entire CRISPR system is composed of two major
NU
components such as Cas9, which is a double stranded DNA nuclease and a personalised cofactor
(synthetic guide RNA) that could selectively target genes to induce DNA breakage. Bacillus subtilis
ATCC 6051 is an active strain involved in the production of industrial enzymes with its outstanding
MA

ability in secreting proteins. The major hurdles experienced by the researchers on using this strain are
insignificant transformation efficiency. Under such positions, CRISPR/Cas9 system was preferred by
Zhang et.al [73]. Future studies on protease production can think of using CRISPR/Cas9 system to
D

enhance gene editing, thereby improvising gene expression of industrially important proteases.
E
PT

A lot of computer based software are user friendly to perform designing the synthetic DNA segments.
Huge number of codon optimization algorithms are available, through which the DNA construct can be
optimized for high level of expression of protein in selected host organism. Gene Designer is a software
CE

available for molecular biologists to expand research in synthetic biology [74]. Another important thrust
area in sequencing related segment is database mining. Metagenomics is a perfect tool through which
AC

DNA could be isolated from environmental micro-organisms and the sequence information is
processed. The technique is followed by screening of open reading frames from the isolated sample in
order to identify any novel genes that could code for a novel enzyme [75]. Through metagenomics
screening, a lot many enzymes have been identified and isolated with novel bio-catalytic activity. There
was a report stating that, by using metagenomics technology a novel protease enzyme had been
identified by Waschkowitz [76]. Developing techniques like metaproteomics and metatranscriptomics
are the potential fields on which researchers could put more attention in order to find a novel enzyme
or a novel source.
ACCEPTED MANUSCRIPT

Compared to older days, more number of enzyme engineering techniques have emerged to improvise
the yield and the performance of the protein fraction. A detailed description about these factors were
discussed by Lutz and Stefan [77]. A few among them are phage display [78] and cell free system for
enzyme production [79]. Manufacturing enzymes is a sustainable process that are highly associated
with synthesis of chemicals. A noteworthy effort is being made every day to explore new enzymes and
also in enhancing the efficiency of existing enzymes [80]. One of the major hiccups in analysing
enzymatic reactions are, the substrate and products are not being distinguished separately by optical

PT
spectroscopy. To overcome such situations, researchers are following separation based assays so that
the product and the substrate are detected individually. Capillary electrophoresis has been proved to be

RI
a better separation technique than chromatography on performing differentiation of substrate and
product [81]. Though, there are several upcoming and existing techniques in enzyme production and

SC
usage, the demand for enzyme still persists in food and pharmaceutical sector. The enzyme synthesized
so far, could be insufficient to meet the need of the large growing population, and the impact of the
NU
enzyme market on developing countries like India is enormous [82].

The last decade has witnessed an alarming achievements in science and technology with extensive
MA

potential of nanotechnology. Their convoluted characters like high surface-to-volume ratio, stability,
higher adsorption and catalysis efficiency make them a suitable platform for achieving target product
various disciplines in science. In the second half of 1980s, nanotechnology has been applied in enzyme
D

production and characterisation. Chiefly, the nanoparticles serve as a matrix for immobilizing the
E

enzyme biomolecules because of their higher surface area and strong adsorption forces [82].
PT

Immobilisation, on the whole improves the stability of the enzyme and also safeguards protein from
unfolding. Another very important criteria of using immobilised enzymes are, the enzyme molecules
behave as free enzymes and exposes Brownian movement in aqueous environment, averts the
CE

aggregation of enzymatic biomolecules thereby enriching enzyme activity. Numerous types such as
metal oxide nanoparticles, polymeric nanoparticles, metal nanoparticles and magnetic nanoparticles
AC

serves as a base support material with extraordinary properties as an enzyme protectant. The enzyme
molecule is attached to the nanoparticles by any of the four mechanisms; a) Conjugation to the surface
of the nanoparticle directly; b) Adsorption by electrostatic forces; c) Affinity oriented protein
conjugation and d) Surface attachment to reformed nanoparticles by covalence. The detailed description
of the processes was mentioned by Razi Ahmad and Meryam Sardar [75].

5. Selected applications of proteases

5.1 Detergent Industry


ACCEPTED MANUSCRIPT

The statistics states that nearly 25-30% of the commercial enzymes are the key ingredients in detergent
making. Amylases and proteases are being extensively used in detergent synthesis process to remove
starch and protein based dirt. In 1913, first enzyme based detergent namely Brunus was introduced and
at the present scenario Novozymes and Genencor are the major suppliers of detergent enzymes [83].
The mode of action of cleaning the stains in the cloth by the detergent is depicted in the figure 3.

The Several alkaline proteases such as serine alkaline protease from Micromonospora chaiyaphumensis
are extremely heat stable and increased thermoactivity was observed in presence of calcium ions [84].

PT
These properties make them ideally fit for applications in detergent making. More recently, intracellular
serine protease from Bacillus megaterium was very efficient in hydrolysing casein and haemoglobin

RI
[85]. Keratinolytic proteases are recently involved, thereby breaking down the major bottlenecks in
protease based detergent production and marketing [86]. The researchers are focusing on developing

SC
detergent with a combination of enzymes to improve the performance. A study states that destaining
ability was high when mannanase and protease are coproduced and used together in a detergent powder
NU
[87]. Generally, protease enzyme powder would be produced and mixed with dry ingredients in
detergent making process. Heat sensitive ingredients are added just prior to spray drying process. The
outline of detergent making is illustrated in figure 4.
MA

5.2 Leather industry


D

Dehairing is an important process in leather industry and usage of chemicals such as sodium sulphide
E

leads various pollution related issues. The protease based dehairing process is eco-friendly and exhibit
PT

favourable outcomes such as strength and quality of the product. Preceding the decision of the protease
for dehairing, a through screening must be done to obtain substrate specific enzyme thereby not
degrading other substrates [88]. The usage of various protease at different steps of tanning operations
CE

are depicted in figure 5.


AC

5.3 Food industry

To hydrolyse the caseins, proteases are added to milk in cheese production process. There has been
serious hunt to find an alternative for usage of rennet in cheese making due to various limitations. Plant
based proteases are identified as a potential source with having potential as that of rennet [89]. The
enzymes used in cheese production recently and their sources are shown in table 5 and the methodology
is shown in the figure 6.

Table 5 Recent proteases in cheese making


ACCEPTED MANUSCRIPT

Protease type Source Reference


Aspartic protease Penicillium brevicompactum [90]
Serine protease Foeniculum vulgare [91]
Preprogaline (aspartic protease) Pichia pastoris [92]
Protease Bacillus species P45 [93]
Aspartic protease Withania coagulans [94]
Aspartic protease Cynara scolymus [95]
Aspartic protease Aspergillus foetidus [96]

PT
Protease Bacillus stearothermophilus [97]
Alkaline protease Solanum melongena [1]

RI
Cysteine protease (Porcerain B) Dregea sinensis [98]

SC
The most important aspect for meats from beef, pork and lamb is its tenderness. Meats are stored at
NU
refrigerated condition and while aging, the proteases present in muscle would break myofibrils thereby
tenderizing it. Cysteine protease calpain 1 (calcium dependant) is an enzyme of prime importance for
softening the meat [99]. A new study explains that the gene coding for protease in Rhizomucor miehei
MA

(Rmpro-A) is expressed in Pichia pastoris and the product exhibited a high range of tenderization in
pork [100]. Usually, papain served as a potential enzyme for softening the meat over a period of time
and the recent finding shows that Rmpro-A, at a very low concentration exhibits tenderization much
D

better than papain. Actinidin is a cysteine protease from chinense kiwifruit and showed improved
E

tenderness properties in rabbit and pork muscles [101]. A new identification named proline
PT

iminopeptidase from lactic acid bacteria is a serine protease that can significantly degrade collagen and
tenderize the meat [102]. Buffaloes are exclusively slaughtered by humankind for meat. Plant
proteolytic enzymes from Cucumis trigonus and Zingiber officinale are nowadays more efficient in
CE

tenderizing buffalo meat than papain [103].


AC

The baking industries’ prime research focus is on degrading gluten, in order to improve the quality of
wheat that is involved in dough preparation. A wide variety of plant and microbe based proteases are
utilised in gluten degradation. Neutral proteases are perfect fit for the baking and brewing applications.
Neutrase is a metalloprotease from Bacillus amyloliquefaciens and it is very potent in hydrolysing meat
and gelatin. Neutrase plays a vital role in modification of dough processing for preparation of biscuits
and crackers [104]. Renzetti and Arendt stated that degradation by proteases could be very beneficial
in enhancing the breadmaking ability of brown rice flour [105].

The non-hydrolysed protein content in other milk formulas are allergic and also quite hard to digest by
children. Proteases are very useful in preparing hydrolysed formula milk for infants [106]. More focus
ACCEPTED MANUSCRIPT

is being thrown at the production of gluten free beer at the brewing industry. A study shows that
recombinant proline specific prolyl endopeptidase from Sphaerobacter thermophiles is very efficient
in producing gluten free beer. The isolated recombinant protein was added to barley during malting
process resulting in high quality gluten free beer [107].

5.4 Therapeutic applications of Protease

Non-invasive therapeutic procedures are largely evolving in treatment of human diseases and disorders.

PT
A broad spectrum of medical conditions is being treated through enzymatic degradation of substrate.
The success rate of such treatment is enormous because of specificity of particular enzyme towards

RI
targeted substrate thereby not harming the surrounding cells. Some of the enzymes are highly selective
and specific even at a very low concentration. Collagenase group of enzymes are matrix

SC
metalloproteinases that can treat conditions such as Dupuytren’s disease, Peyronie’s disease, wound
healing, burns, glaucoma, intervertebral disc herniation, debridement, degradation of human retained
NU
placenta, cartilage repair, nipple pain, keloid, vitrectomy, cellulite, chronic total occlusions, gene
delivery and uterine fibroid. These medical conditions and the responsible collagen type used for
treatments are elaborated in detail by Hamzeh Alipour et al [108].
MA

Enzymes are also formulated as topical applications. Alkaline proteases from Bacillus subtilis acts as
efficient transdermal drug delivery system for dermatological applications [109]. Cysteine proteases are
D

best in being a validated target for treating ailments. Cathepsins and calpains are pharmaceutically more
E

important cysteine proteases. Cysteine proteases glucoprotein purified from Zingiber montanum proved
PT

to be a very potent anti-oxidant [110]. A recent study shows that cysteine protease from Sesbania
grandiflora is very effective in exhibiting haemostatic and fibrinogenolytic activity [111].
Cardiometabolic diseases are highly treated with matrix metalloproteinases. Their mode of action is
CE

through modifying extracellular matrix components like collagen, gelatin and fibronectin [112]. The
overall therapeutic application of proteases is illustrated in figure 7.
AC

5.5 Protease Formulations


Researchers are focusing on preparing the protease enzymes into human friendly formulations for its
utmost usage in various application sectors. A few examples are discussed here. As proteases are very
dynamic in action, the enzyme is recommended as a skin care formulation for utility. Desquamation is
an important process that helps in maintaining the health of a skin. Physiological desquamation along
with functions of epidermal barrier, are completely dependent on water content of the body and also on
the presence of protease biomolecules and their inhibitors [113].
ACCEPTED MANUSCRIPT

Historically proteases have been used in the treatment of skin ulcers as reported by Ramundo and Gray
[114]. Recent advancements suggest that proteases could be used as a skin cleanser and also as a leave
on application in order to degrade the skin dirt which could be removed off from the skin by the
detergent component of the formulation [115]. Apart from skin care, proteases are being made into
formulations in order to improve the digestive ability of humans. An invention in United States patent
publications reports that, the usage of fungal protease along with effective excipients could act as a
health supplement for improving digestion and also equally active in raising pH level of blood which
has its own health benefits [116]. Beside skin care and digestive formulation applications, there are

PT
emerging protease based tooth-paste [117], protease capsules for wellness [118] and protease based
detergents [119]. Currently, with advanced techniques emerging in, researchers are emphasizing on

RI
synthesizing more formulations with protease as an active ingredient in all industries.

SC
6. A view on the statistics
Protease has been a great attraction to the researchers because of its widespread application potential.
NU
Over the period, huge number of research publications had been published in reputed journals to
establish the efficiency of proteases. The figure 8 and figure 9 shows the statistical data of proteases
retrieved from Scopus database. It includes year-wise publication on proteases for the past ten years;
MA

protease sources and their applications in percentages. The data showed that for the period of ten years
the research interest on proteases had kept increasing without a drop. It motivates the future researchers
and scientists to do study on proteases and produce more commercially viable products for the society.
D

On analysing the patent scenario, there are around 215 patent publication over ten years, existing in
E

India among which 21 were granted. The mentioned data are obtained from Indian Patent Advance
Search System – In PASS, Indian Patent Database. Globally, 5700 patent publications are available
PT

from 2008 to 2018 (source: worldwide database search – EPO). As per the World Bank Data, the Indian
Annual GDP (Gross Domestic Product) has been improved from 6.687 % in 2017 to 7.327 % in 2018
CE

(https://data.worldbank.org/country/india). The World Bank Forecasts Annual GDP for India in 2019
is 7.536%. It is a well-known fact that the food and beverages industry contributes a lot more to GDP
AC

growth. In order to achieve the increase in food sector product development, researchers should focus
on upbringing high yielding varieties through gene manipulation and also in synthesis of low cost and
high value products through economical downstream processing systems, which in turn could be
accessible and affordable to all the people in the society. On analysing the global scenario, America is
the leading consumer of enzymes and while considering Asian continent China is the larger supplier of
enzymes for various purposes followed by Japan. Certain surveys states that Western Europe would be
largest enzyme producer within a period of five to ten years [113] .

As we are in the concluding segment, it is more vital to think about future prospects of the protease
enzyme in medicines production sector. On analysing various reports, research publications and articles
ACCEPTED MANUSCRIPT

it was well understood that the downstream portion of the protease production involves enormous
money to be spent on it. The future research should be morally focused on developing economic
purification strategy of industrial proteases. The researchers would bring up on efficient substitutes to
column packing materials thereby cutting down the investment on downstream processing. To meet up
the affordable medical need of the people, scientists must explore on emergent pharmaceutical
applications of the protease enzyme. The finding of novel protease with economical downstream
processing costs and targeted therapeutical application, it is greatly achievable to serve the human
community to come over the medical struggles.

PT
7. Conclusion

RI
Proteases are creating a benchmark in every field with their potency and it is one of the most rapidly

SC
expanding area of exploration. Large numbers of researchers are focusing on engineering protease
producing genes for enhancing the quality and quantity of the product. A broad spectrum of human
NU
medical conditions is treated by protease enzyme and the success rate is enormous. In food industry,
the applications of protease are massive and measures are being taken to enhance the safety and value
of the food. Leather product manufacturers are purely dependant on enzyme processing for de-hairing
MA

in order to minimise the pollution created by the synthetic chemicals. The uncontrolled proteolysis
might be harmful in some ways but, with advancements in science and technology the methods to use
protease has been growing widespread thereby benefiting humankind.
E D

8. References
PT

[1] A. Kumari, B. Kaur, R. Srivastava, R.S. Sangwan, Isolation and immobilization of alkaline
protease on mesoporous silica and mesoporous ZSM-5 zeolite materials for improved catalytic
CE

properties, Biochem. Biophys. Reports. 2 (2015) 108–114. doi:10.1016/j.bbrep.2015.05.009.


[2] R. Ramachandran, F. Noorbakhsh, K. Defea, M.D. Hollenberg, Targeting proteinase-activated
AC

receptors: Therapeutic potential and challenges, Nat. Rev. Drug Discov. (2012).
doi:10.1038/nrd3615.
[3] K. Kato, A. Hara, T. Kuno, N. Kitaori, Z. Huilan, H. Mori, M. Toida, T. Shibata, Matrix
metalloproteinases 2 and 9 in oral squamous cell carcinomas: manifestation and localization of
their activity, J. Cancer Res. Clin. Oncol. 131 (2005) 340–346. doi:10.1007/s00432-004-0654-
8.
[4] F. Stegmeier, M. Rape, V.M. Draviam, G. Nalepa, M.E. Sowa, X.L. Ang, E.R. McDonald III,
M.Z. Li, G.J. Hannon, P.K. Sorger, M.W. Kirschner, J.W. Harper, S.J. Elledge, Anaphase
initiation is regulated by antagonistic ubiquitination and deubiquitination activities, Nature.
446 (2007) 876–881. doi:10.1038/nature05694.
ACCEPTED MANUSCRIPT

[5] C.G. Kumar, H. Takagi, Microbial alkaline proteases: from a bioindustrial viewpoint.,
Biotechnol. Adv. 17 (1999) 561–94.
[6] Enzyme Nomenclature. Recommendations 1992. Supplement: corrections and additions, Eur.
J. Biochem. 223 (1994) 1–5. doi:10.1111/j.1432-1033.1994.tb18960.x.
[7] S. Guleria, A. Walia, A. Chauhan, C.K. Shirkot, Molecular characterization of alkaline
protease of Bacillus amyloliquefaciens SP1 involved in biocontrol of Fusarium oxysporum,
Int. J. Food Microbiol. 232 (2016) 134–143. doi:10.1016/j.ijfoodmicro.2016.05.030.
[8] S.K. Agarwal, Proteases cathepsins — A view, Biochem. Educ. 18 (1990) 67–72.

PT
doi:10.1016/0307-4412(90)90176-O.
[9] J.K. McDonald, An overview of protease specificity and catalytic mechanisms: aspects related

RI
to nomenclature and classification, Histochem. J. 17 (1985) 773–785.
doi:10.1007/BF01003313.

SC
[10] T.J. Motyan JA, Toth F, Research Applications of Proteolytic Enzymes in Molecular Biology,
Biomolecules. 3 (2013) 923–942.
NU
[11] M.B. Rao, A.M. Tanksale, M.S. Ghatge, V. V Deshpande, Molecular and biotechnological
aspects of microbial proteases., Microbiol. Mol. Biol. Rev. 62 (1998) 597–635.
[12] A. Taylor, Aminopeptidases: structure and function., FASEB J. 7 (1993) 290–8.
MA

[13] W.T. Lowther, B.W. Matthews, Structure and function of the methionine aminopeptidases.,
Biochim. Biophys. Acta. 1477 (2000) 157–67.
http://www.ncbi.nlm.nih.gov/pubmed/10708856.
D

[14] N. Rani, A. Aichem, G. Schmidtke, S.G. Kreft, M. Groettrup, FAT10 and NUB1L bind to the
E

VWA domain of Rpn10 and Rpn1 to enable proteasome-mediated proteolysis., Nat. Commun.
PT

3 (2012) 749. doi:10.1038/ncomms1752.


[15] R. Sawant, S. Nagendran, Protease : an Enzyme With Multiple Industrial Applications, World
J. Pharm. Pharm. Sci. 3 (2014) 568–579.
CE

[16] A. Laskar, A. Chatterjee, S. Chatterjee, E.J. Rodger, Three-Dimensional Molecular Modeling


of a Diverse Range of SC Clan Serine Proteases, Mol. Biol. Int. 2012 (2012) 1–9.
AC

doi:10.1155/2012/580965.
[17] E. Di Cera, Serine proteases, IUBMB Life. 61 (2009) 510–515. doi:10.1002/iub.186.
[18] C.M. Antão, F.X. Malcata, Plant serine proteases: biochemical, physiological and molecular
features., Plant Physiol. Biochem. PPB. 43 (2005) 637–50. doi:10.1016/j.plaphy.2005.05.001.
[19] P.B. Szecsi, The aspartic proteases., Scand. J. Clin. Lab. Invest. Suppl. 210 (1992) 5–22.
[20] M. Siklos, M. BenAissa, G.R.J. Thatcher, Cysteine proteases as therapeutic targets: does
selectivity matter? A systematic review of calpain and cathepsin inhibitors, Acta Pharm. Sin.
B. 5 (2015) 506–519. doi:10.1016/j.apsb.2015.08.001.
[21] S. Rani, Kirti & Rana, Rachita & Datt, Review on latest overview of proteases., Int. J. Curr.
Life Sci. 2 (2012) 12–18.
ACCEPTED MANUSCRIPT

[22] T.T. Baird, W.D. Wright, C.S. Craik, Conversion of trypsin to a functional threonine protease.,
Protein Sci. 15 (2006) 1229–1238. doi:10.1110/ps.062179006.
[23] A.H. Sims, N.S. Dunn-Coleman, G.D. Robson, S.G. Oliver, Glutamic protease distribution is
limited to filamentous fungi, FEMS Microbiol. Lett. 239 (2004) 95–101.
doi:10.1016/j.femsle.2004.08.023.
[24] M.B. Rao, A.M. Tanksale, M.S. Ghatge, V. V. Deshpande, Molecular and Biotechnological
Aspects of Microbial Proteases, Microbiol. Mol. Biol. Rev. 62 (1998) 597–635.
doi:papers2://publication/uuid/E58ABF6D-8C97-4209-810D-A452EE30B2CD.

PT
[25] R.P. Yadav, A.K. Patel, M. V. Jagannadham, Purification and biochemical characterization of
a chymotrypsin-like serine protease from Euphorbia neriifolia Linn., Process Biochem. 46

RI
(2011) 1654–1662. doi:10.1016/j.procbio.2011.05.013.
[26] S.B. Badgujar, R.T. Mahajan, Proteolytic enzymes of some laticiferous plants belonging to

SC
Khandesh region of Maharashtra, India., J. Pharm. Res. 2 (2009) 1434–1437.
http://jpronline.info/article/view/562/482.
NU
[27] L.P. Moro, H. Cabral, D.N. Okamoto, I. Hirata, M.A. Juliano, L. Juliano, G.O. Bonilla-
Rodriguez, Characterization, subsite mapping and N-terminal sequence of miliin, a serine-
protease isolated from the latex of Euphorbia milii, Process Biochem. 48 (2013) 633–637.
MA

doi:10.1016/j.procbio.2013.02.017.
[28] L.P. Moro, M.T. Murakami, H. Cabral, A. Vidotto, E.H. Tajara, R.K. Arni, L. Juliano, G.O.
Bonilla-Rodriguez, Purification, biochemical and functional characterization of miliin, a new
D

thiol-dependent serine protease isolated from the latex of Euphorbia milii., Protein Pept. Lett.
E

15 (2008) 221–227. doi:10.2174/092986608785133744.


PT

[29] C.T. Butts, X. Zhang, J.E. Kelly, K.W. Roskamp, M.H. Unhelkar, J.A. Freites, S. Tahir, R.W.
Martin, Sequence comparison, molecular modeling, and network analysis predict structural
diversity in cysteine proteases from the Cape sundew, Drosera capensis, Comput. Struct.
CE

Biotechnol. J. 14 (2016) 271–282. doi:10.1016/j.csbj.2016.05.003.


[30] R. Tomar, R. Kumar, M. V. Jagannadham, A stable serine protease, wrightin, from the latex of
AC

the plant Wrightia tinctoria (Roxb.) R. Br.: Purification and biochemical properties, J. Agric.
Food Chem. 56 (2008) 1479–1487. doi:10.1021/jf0726536.
[31] E. Amri, F. Mamboya, Papain, a plant enzyme of biological importance: A review, Am. J.
Biochem. Biotechnol. 8 (2012) 99–104. doi:10.3844/ajbbsp.2012.99.104.
[32] A.K. Patel, V.K. Singh, M. V. Jagannadham, Carnein, a serine protease from noxious plant
weed Ipomoea carnea (Morning glory), J. Agric. Food Chem. 55 (2007) 5809–5818.
doi:10.1021/jf063700h.
[33] M.G. Guevara, P. Veríssimo, E. Pires, C. Faro, G.R. Daleo, Potato aspartic proteases:
Induction, antimicrobial activity and substrate specificity, J. Plant Pathol. 86 (2004) 233–238.
doi:10.2307/41992430.
ACCEPTED MANUSCRIPT

[34] L.R. Singh, T.P. Devi, S.K. Devi, Purification and Characterization of a Pineapple Crown Leaf
Thiol Protease, Prep. Biochem. Biotechnol. 34 (2004) 25–43. doi:10.1081/PB-120027111.
[35] . S.T.E.-S., Purification and Characterization of Raphanin, A Neutral Protease, from Raphanus
sativus Leaves, Pakistan J. Biol. Sci. 4 (2001) 564–568. doi:10.3923/pjbs.2001.564.568.
[36] W.B. Jiang, A. Lers, E. Lomaniec, N. Aharoni, Senescence-related serine protease in parsley,
Phytochemistry. 50 (1998) 377–382. doi:10.1016/S0031-9422(98)00546-9.
[37] B. Nieri, S. Canino, R. Versace, A. Alpi, Purification and characterization of an endoprotease
from alfalfa senescent leaves, Phytochemistry. 49 (1998) 643–649. doi:10.1016/S0031-

PT
9422(98)00280-5.
[38] J. Parkash, S. Kashyap, S. Kirti, A.K. Singh, S. Dutt, Cathepsin B cysteine protease gene is

RI
upregulated during leaf senescence and exhibits differential expression behavior in response to
phytohormones in Picrorhiza kurrooa Royle ex Benth, Plant Gene. 3 (2015) 11–19.

SC
doi:10.1016/j.plgene.2015.07.001.
[39] M. Diaz-Mendoza, B. Velasco-Arroyo, M.E. Santamaria, P. Gonz�lez-Melendi, M.
NU
Martinez, I. Diaz, Plant senescence and proteolysis: Two processes with one destiny, Genet.
Mol. Biol. 39 (2016) 329–338. doi:10.1590/1678-4685-GMB-2016-0015.
[40] Z.I.M. Arshad, A. Amid, F. Yusof, I. Jaswir, K. Ahmad, S.P. Loke, Bromelain: An overview
MA

of industrial application and purification strategies, Appl. Microbiol. Biotechnol. 98 (2014)


7283–7297. doi:10.1007/s00253-014-5889-y.
[41] A. Muhlia-Almazán, A. Sánchez-Paz, F.L. García-Carreño, Invertebrate trypsins: A review, J.
D

Comp. Physiol. B Biochem. Syst. Environ. Physiol. 178 (2008) 655–672. doi:10.1007/s00360-
E

008-0263-y.
PT

[42] D.M. Blow, Structure and Mechanism of Chymotrypsin, Acc. Chem. Res. 9 (1976) 145–152.
doi:10.1021/ar50100a004.
[43] L. Zhao, S. M Budge, A. E Ghaly, M. S Brooks, D. Dave, Extraction, Purification and
CE

Characterization of Fish Pepsin: A Critical Review, J. Food Process. Technol. 2 (2011).


doi:10.4172/2157-7110.1000126.
AC

[44] A.A. Shinde, F.K. Shaikh, M. V. Padul, M.S. Kachole, Bacillus subtillis RTSBA6 6.00, a new
strain isolated from gut of Helicoverpa armigera (Lepidoptera: Noctuidae) produces
chymotrypsin-like proteases, Saudi J. Biol. Sci. 19 (2012) 317–323.
doi:10.1016/j.sjbs.2012.03.001.
[45] P. Vijayaraghavan, S. Lazarus, S.G.P. Vincent, De-hairing protease production by an isolated
Bacillus cereus strain AT under solid-state fermentation using cow dung: Biosynthesis and
properties, Saudi J. Biol. Sci. 21 (2014) 27–34. doi:10.1016/j.sjbs.2013.04.010.
[46] C. Suganthi, A. Mageswari, S. Karthikeyan, M. Anbalagan, A. Sivakumar, K.M. Gothandam,
Screening and optimization of protease production from a halotolerant Bacillus licheniformis
isolated from saltern sediments, J. Genet. Eng. Biotechnol. 11 (2013) 47–52.
ACCEPTED MANUSCRIPT

doi:10.1016/j.jgeb.2013.02.002.
[47] M.M.S. Asker, M.G. Mahmoud, K. El Shebwy, M.S. Abd el Aziz, Purification and
characterization of two thermostable protease fractions from Bacillus megaterium, J. Genet.
Eng. Biotechnol. 11 (2013) 103–109. doi:10.1016/j.jgeb.2013.08.001.
[48] M. Venugopal, A. V. Saramma, An alkaline protease from Bacillus circulans BM15, newly
isolated from a mangrove station: Characterization and application in laundry detergent
formulations, Indian J. Microbiol. 47 (2007) 298–303. doi:10.1007/s12088-007-0055-1.
[49] B. Johnvesly, B.R. Manjunath, G.R. Naik, Pigeon pea waste as a novel, inexpensive, substrate

PT
for production of a thermostable alkaline protease from thermoalkalophilic Bacillus sp. JB-99,
Bioresour. Technol. 82 (2002) 61–64. doi:10.1016/S0960-8524(01)00147-X.

RI
[50] S. Sivaprakasam, B. Dhandapani, S. Mahadevan, Optimization studies on production of a salt-
tolerant protease from Pseudomonas aeruginosa strain BC1 and its application on tannery

SC
saline wastewater treatment, Brazilian J. Microbiol. 42 (2011) 1506–1515. doi:10.1590/S1517-
83822011000400038.
NU
[51] E. Domann, M. Leimeisterwachter, W. Goebel, T. Chakraborty, MOLECULAR-CLONING,
SEQUENCING, AND IDENTIFICATION OF A METALLOPROTEASE GENE FROM
LISTERIA-MONOCYTOGENES THAT IS SPECIES-SPECIFIC AND PHYSICALLY
MA

LINKED TO THE LISTERIOLYSIN GENE, Infect. Immun. 59 (1991) 65–72.


[52] N. Shafee, C.C. Tan, S. Mahamad, R.N.Z.A. Rahman, M. Basri, A.B. Salleh, Nutritional
factors affecting organic solvent-tolerant alkaline protease production by a new Bacillus cereus
D

strain 146, Ann. Microbiol. 56 (2006) 29–34. doi:10.1007/BF03174966.


E

[53] D.C. Brenda, K. Dnyanada, K.D. Santosh, Partial purification and characterization of
PT

metalloprotease of halotolerant alkaliphilic bacterium Bacillus cereus from coastal sediment of


Goa, India, African J. Biotechnol. 12 (2013) 4905–4914. doi:10.5897/AJB2013.12808.
[54] X.G. Chen, O. Stabnikova, J.H. Tay, J.Y. Wang, S.T.L. Tay, Thermoactive extracellular
CE

proteases of Geobacillus caldoproteolyticus, sp. nov., from sewage sludge, Extremophiles. 8


(2004) 489–498. doi:10.1007/s00792-004-0412-5.
AC

[55] M. Arulmani, K. Aparanjini, K. Vasanthi, P. Arumugam, M. Arivuchelvi, P.T. Kalaichelvan,


Purification and partial characterization of serine protease from thermostable alkalophilic
Bacillus laterosporus-AK1, World J. Microbiol. Biotechnol. 23 (2007) 475–481.
doi:10.1007/s11274-006-9249-7.
[56] K. Kalaiarasi, P.U. Sunitha, Optimization of alkaline protease production from Pseudomonas
fluorescens isolated from meat waste contaminated soil, African J. Biotechnol. 8 (2009) 7035–
7041. http://www.academicjournals.org/AJB.
[57] K.M. Sharma, R. Kumar, S. Panwar, A. Kumar, Microbial alkaline proteases: Optimization of
production parameters and their properties, J. Genet. Eng. Biotechnol. 15 (2017) 115–126.
doi:10.1016/j.jgeb.2017.02.001.
ACCEPTED MANUSCRIPT

[58] W.H. Chu, Optimization of extracellular alkaline protease production from species of Bacillus,
J. Ind. Microbiol. Biotechnol. 34 (2007) 241–245. doi:10.1007/s10295-006-0192-2.
[59] S. Gaur, S. Agrahari, N. Wadhwa, Purification of Protease from Pseudomonas thermaerum
GW1 Isolated from Poultry Waste Site., Open Microbiol. J. 4 (2010) 67–74.
doi:10.2174/1874285801004010067.
[60] L. Cui, Q.H. Liu, H.X. Wang, T.B. Ng, An alkaline protease from fresh fruiting bodies of the
edible mushroom Pleurotus citrinopileatus, Appl. Microbiol. Biotechnol. 75 (2007) 81–85.
doi:10.1007/s00253-006-0801-z.

PT
[61] N.W. Hsiao, Y. Chen, Y.C. Kuan, Y.C. Lee, S.K. Lee, H.H. Chan, C.H. Kao, Purification and
characterization of an aspartic protease from the Rhizopus Oryzae protease extract, peptidase

RI
R, Electron. J. Biotechnol. 17 (2014) 89–94. doi:10.1016/j.ejbt.2014.02.002.
[62] A.R.G. Machado, M.F.S. Teixeira, L. de Souza Kirsch, M. da C.L. Campelo, I.M. de Aguiar

SC
Oliveira, Nutritional value and proteases of Lentinus citrinus produced by solid state
fermentation of lignocellulosic waste from tropical region, Saudi J. Biol. Sci. 23 (2016) 621–
NU
627. doi:10.1016/j.sjbs.2015.07.002.
[63] Khademi F, Abachi S, Mortazavi A, Ehsani M A, Tabatabaei M R, Optimization of Fungal
Rennet Production by local isolate of Rhizomucor nainitalensis Under Solid Substrate
MA

Fermentation system, n.d.


[64] S. Chellappan, C. Jasmin, S.M. Basheer, K.K. Elyas, S.G. Bhat, M. Chandrasekaran,
Production, Purification and Partial Characterization of a Novel Protease from Marine
D

<i>Engyodontium album<i> BTMFS10 under Solid State Fermentation, Process Biochem. 41


E

(2006) 956–961. doi:10.1016/j.procbio.2005.10.017.


PT

[65] D. Agrawal, P. Patidar, T. Banerjee, S. Patil, Alkaline protease production by a soil isolate of
Beauveria felina under SSF condition: Parameter optimization and application to soy protein
hydrolysis, Process Biochem. 40 (2005) 1131–1136. doi:10.1016/j.procbio.2004.03.006.
CE

[66] R. Sindhu, G.N. Suprabha, S. Shashidhar, Optimization of process parameters for the
production of alkaline protease from Penicillium godlewskii SBSS 25 and its application in
AC

detergent industry, African J. Microbiol. Res. 3 (2009) 515–522.


[67] S. Almas, A. Hameed, D. Shelly, P. Mohan, Purification and characterization of a novel
protease from Bacillus strain SAL1, African J. Biotechnol. 8 (2009) 3603–3609.
doi:10.5897/AJB09.731.
[68] M. Hajji, S. Kanoun, M. Nasri, N. Gharsallah, Purification and characterization of an alkaline
serine-protease produced by a new isolated Aspergillus clavatus ES1, Process Biochem. 42
(2007) 791–797. doi:10.1016/j.procbio.2007.01.011.
[69] V. Benluvankar, G.R. Jebapriya, J.J. Gnanadoss, PROTEASE PRODUCTION BY
PENICILLIUM SP. LCJ228 UNDER SOLID STATE FERMENTATION USING
GROUNDNUT OILCAKE AS SUBSTRATE, (n.d.).
ACCEPTED MANUSCRIPT

http://www.ijlpr.com/admin/php/uploads/222_pdf.pdf (accessed October 3, 2018).


[70] P. Mitra, P.K. Chakrabartty, An extracellular protease with depilation activity from
Streptomyces nogalator, J. Sci. Ind. Res. (India). 64 (2005) 978–983.
[71] B.K. Bajaj, P. Sharma, An alkali-thermotolerant extracellular protease from a newly isolated,
N. Biotechnol. 28 (2011) 725–732. doi:10.1016/j.nbt.2011.01.001.
[72] V. Martínez, I. Lauritsen, T. Hobel, S. Li, A.T. Nielsen, M.H.H. Nørholm, CRISPR/Cas9-
based genome editing for simultaneous interference with gene expression and protein
stability., Nucleic Acids Res. 45 (2017) e171. doi:10.1093/nar/gkx797.

PT
[73] K. Zhang, X. Duan, J. Wu, Multigene disruption in undomesticated Bacillus subtilis ATCC
6051a using the CRISPR/Cas9 system., Sci. Rep. 6 (2016) 27943. doi:10.1038/srep27943.

RI
[74] A. Villalobos, J.E. Ness, C. Gustafsson, J. Minshull, S. Govindarajan, Gene Designer: a
synthetic biology tool for constructing artificial DNA segments, BMC Bioinformatics. 7

SC
(2006) 285. doi:10.1186/1471-2105-7-285.
[75] R.A. Meryam Sardar, M. Sardar, Enzyme Immobilization: An Overview on Nanoparticles as
NU
Immobilization Matrix, Biochem. Anal. Biochem. 4 (2015) 1–8. doi:10.4172/2161-
1009.1000178.
[76] T. Waschkowitz, S. Rockstroh, R. Daniel, Isolation and characterization of metalloproteases
MA

with a novel domain structure by construction and screening of metagenomic libraries., Appl.
Environ. Microbiol. 75 (2009) 2506–16. doi:10.1128/AEM.02136-08.
[77] S. Lutz, Beyond directed evolution—semi-rational protein engineering and design, Curr. Opin.
D

Biotechnol. 21 (2010) 734–743. doi:10.1016/j.copbio.2010.08.011.


E

[78] J. Löfblom, Bacterial display in combinatorial protein engineering, Biotechnol. J. 6 (2011)


PT

1115–1129. doi:10.1002/biot.201100129.
[79] Y. Ma, D. Münch, T. Schneider, H.-G. Sahl, A. Bouhss, U. Ghoshdastider, J. Wang, V.
Dötsch, X. Wang, F. Bernhard, Preparative Scale Cell-free Production and Quality
CE

Optimization of MraY Homologues in Different Expression Modes, J. Biol. Chem. 286 (2011)
38844–38853. doi:10.1074/jbc.M111.301085.
AC

[80] M.J. Liszka, M.E. Clark, E. Schneider, D.S. Clark, Nature Versus Nurture: Developing
Enzymes That Function Under Extreme Conditions, Annu. Rev. Chem. Biomol. Eng. 3 (2012)
77–102. doi:10.1146/annurev-chembioeng-061010-114239.
[81] S. Gattu, C.L. Crihfield, G. Lu, L. Bwanali, L.M. Veltri, L.A. Holland, Advances in enzyme
substrate analysis with capillary electrophoresis., Methods. 146 (2018) 93–106.
doi:10.1016/j.ymeth.2018.02.005.
[82] R. Singh, M. Kumar, A. Mittal, P.K. Mehta, Microbial enzymes: industrial progress in 21st
century., 3 Biotech. 6 (2016) 174. doi:10.1007/s13205-016-0485-8.
[83] D. Kumar, . S., N. Thakur, R. Verma, T.C. Bhalla, Microbial Proteases and Application as
Laundry Detergent Additive, Res. J. Microbiol. 3 (2008) 661–672.
ACCEPTED MANUSCRIPT

doi:10.3923/jm.2008.661.672.
[84] S. Mhamdi, N. Ktari, S. Hajji, M. Nasri, A. Sellami Kamoun, Alkaline proteases from a newly
isolated Micromonospora chaiyaphumensis S103: Characterization and application as a
detergent additive and for chitin extraction from shrimp shell waste, Int. J. Biol. Macromol. 94
(2017) 415–422. doi:10.1016/j.ijbiomac.2016.10.036.
[85] Y.J. Jeong, S.C. Baek, H. Kim, Cloning and characterization of a novel intracellular serine
protease (IspK) from Bacillus megaterium with a potential additive for detergents, Int. J. Biol.
Macromol. 108 (2018) 808–816. doi:10.1016/j.ijbiomac.2017.10.173.

PT
[86] T. Paul, A. Jana, A.K. Mandal, A. Mandal, P.K. Das Mohpatra, K.C. Mondal, Bacterial
keratinolytic protease, imminent starter for NextGen leather and detergent industries, Sustain.

RI
Chem. Pharm. 3 (2016) 8–22. doi:10.1016/j.scp.2016.01.001.
[87] A. David, P. Singh Chauhan, A. Kumar, S. Angural, D. Kumar, N. Puri, N. Gupta,

SC
Coproduction of protease and mannanase from Bacillus nealsonii PN-11 in solid state
fermentation and their combined application as detergent additives, Int. J. Biol. Macromol. 108
NU
(2018) 1176–1184. doi:10.1016/j.ijbiomac.2017.09.037.
[88] S. Shakilanishi, C. Shanthi, Specificity studies on proteases for dehairing in leather processing
using decorin as model conjugated protein, Int. J. Biol. Macromol. 103 (2017) 1069–1076.
MA

doi:10.1016/j.ijbiomac.2017.05.134.
[89] M.A. Shah, S.A. Mir, M.A. Paray, Plant proteases as milk-clotting enzymes in cheesemaking:
A review, Dairy Sci. Technol. 94 (2014) 5–16. doi:10.1007/s13594-013-0144-3.
D

[90] S. Ozturkoglu-Budak, A. Wiebenga, P.A. Bron, R.P. de Vries, Protease and lipase activities of
E

fungal and bacterial strains derived from an artisanal raw ewe’s milk cheese, Int. J. Food
PT

Microbiol. 237 (2016) 17–27. doi:10.1016/j.ijfoodmicro.2016.08.007.


[91] N. Bey, H. Debbebi, F. Abidi, M.N. Marzouki, A. Ben Salah, The non-edible parts of fennel
(Fœniculum vulgare) as a new milk-clotting protease source, Ind. Crops Prod. 112 (2018) 181–
CE

187. doi:10.1016/j.indcrop.2017.10.050.
[92] L. Feijoo-Siota, J.L.R. Rama, A. Sánchez-Pérez, T.G. Villa, Expression, activation and
AC

processing of a novel plant milk-clotting aspartic protease in Pichia pastoris, J. Biotechnol. 268
(2018) 28–39. doi:10.1016/j.jbiotec.2018.01.006.
[93] A.C. Lemes, Y. Pavón, S. Lazzaroni, S. Rozycki, A. Brandelli, S.J. Kalil, A new milk-clotting
enzyme produced by Bacillus sp. P45 applied in cream cheese development, LWT - Food Sci.
Technol. 66 (2016) 217–224. doi:10.1016/j.lwt.2015.10.038.
[94] M. Salehi, M.R. Aghamaali, R.H. Sajedi, S.M. Asghari, E. Jorjani, Purification and
characterization of a milk-clotting aspartic protease from Withania coagulans fruit, Int. J. Biol.
Macromol. 98 (2017) 847–854. doi:10.1016/j.ijbiomac.2017.02.034.
[95] B.E. Llorente, W.D. Obregón, F.X. Avilés, N.O. Caffini, S. Vairo-Cavalli, Use of artichoke
(Cynara scolymus) flower extract as a substitute for bovine rennet in the manufacture of
ACCEPTED MANUSCRIPT

Gouda-type cheese: Characterization of aspartic proteases, Food Chem. 159 (2014) 55–63.
doi:10.1016/j.foodchem.2014.03.007.
[96] P.M. Souza, G. Werneck, B. Aliakbarian, F. Siqueira, E.X. Ferreira Filho, P. Perego, A.
Converti, P.O. Magalhães, A.P. Junior, Production, purification and characterization of an
aspartic protease from Aspergillus foetidus, Food Chem. Toxicol. 109 (2017) 1103–1110.
doi:10.1016/j.fct.2017.03.055.
[97] S.A. Ahmed, H.R. Wehaidy, O.A. Ibrahim, S. Abd El Ghani, M.A. El-Hofi, Novel milk-
clotting enzyme from Bacillus stearothermophilus as a coagulant in UF-white soft cheese,

PT
Biocatal. Agric. Biotechnol. 7 (2016) 241–249. doi:10.1016/j.bcab.2016.06.011.
[98] Y. Zhang, H. Wang, L. Tao, A. Huang, Milk-clotting mechanism of Dregea sinensis Hemsl.

RI
protease, J. Dairy Sci. 98 (2015) 8445–8453. doi:10.3168/jds.2015-9851.
[99] J.D. Morton, Z.F. Bhat, A. El-Din Ahmed Bekhit, Proteases and Meat Tenderization, in: Ref.

SC
Modul. Food Sci., 2018. doi:10.1016/B978-0-08-100596-5.21663-6.
[100] Q. Sun, F. Chen, F. Geng, Y. Luo, S. Gong, Z. Jiang, A novel aspartic protease from
NU
Rhizomucor miehei expressed in Pichia pastoris and its application on meat tenderization and
preparation of turtle peptides, Food Chem. 245 (2018) 570–577.
doi:10.1016/j.foodchem.2017.10.113.
MA

[101] B. Zhang, Q. Sun, H.J. Liu, S.Z. Li, Z.Q. Jiang, Characterization of actinidin from Chinese
kiwifruit cultivars and its applications in meat tenderization and production of angiotensin I-
converting enzyme (ACE) inhibitory peptides, LWT - Food Sci. Technol. 78 (2017) 1–7.
D

doi:10.1016/j.lwt.2016.12.012.
E

[102] P. Chanalia, D. Gandhi, P. Attri, S. Dhanda, Extraction, purification and characterization of


PT

low molecular weight Proline iminopeptidase from probiotic L. plantarum for meat
tenderization, Int. J. Biol. Macromol. 109 (2018) 651–663.
doi:10.1016/j.ijbiomac.2017.12.092.
CE

[103] B.M. Naveena, S.K. Mendiratta, A.S.R. Anjaneyulu, Tenderization of buffalo meat using plant
proteases from Cucumis trigonus Roxb (Kachri) and Zingiber officinale roscoe (Ginger
AC

rhizome), Meat Sci. 68 (2004) 363–369. doi:10.1016/j.meatsci.2004.04.004.


[104] J. Ehren, B. Móron, E. Martin, M.T. Bethune, G.M. Gray, C. Khosla, A food-grade enzyme
preparation with modest gluten detoxification properties, PLoS One. 4 (2009).
doi:10.1371/journal.pone.0006313.
[105] S. Renzetti, E.K. Arendt, Effect of protease treatment on the baking quality of brown rice
bread: From textural and rheological properties to biochemistry and microstructure, J. Cereal
Sci. 50 (2009) 22–28. doi:10.1016/j.jcs.2009.02.002.
[106] D.M. Fleischer, C. Venter, Y. Vandenplas, Hydrolyzed Formula for Every Infant?, Nestle
Nutr. Inst. Workshop Ser. 86 (2016) 51–65. doi:10.1159/000442956.
[107] R. Shetty, M. Vestergaard, F. Jessen, P. Hägglund, V. Knorr, P. Koehler, H.S. Prakash, T.J.
ACCEPTED MANUSCRIPT

Hobley, Discovery, cloning and characterisation of proline specific prolyl endopeptidase, a


gluten degrading thermo-stable enzyme from Sphaerobacter thermophiles, Enzyme Microb.
Technol. 107 (2017) 57–63. doi:10.1016/j.enzmictec.2017.08.002.
[108] H. Alipour, A. Raz, S. Zakeri, N. Dinparast Djadid, Therapeutic applications of collagenase
(metalloproteases): A review, Asian Pac. J. Trop. Biomed. 6 (2016) 975–981.
doi:10.1016/J.APJTB.2016.07.017.
[109] M.I. Nounou, T.I. Zaghloul, N.A. Ahmed, A.A. Eid, L.K. El-Khordagui, Skin permeability
enhancement by Bacillus subtilis alkaline protease: Application to transdermal drug delivery,

PT
Int. J. Pharm. 529 (2017) 423–432. doi:10.1016/j.ijpharm.2017.06.057.
[110] K. Jamir, K. Seshagirirao, Purification, biochemical characterization and antioxidant property

RI
of ZCPG, a cysteine protease from Zingiber montanum rhizome, Int. J. Biol. Macromol. 106
(2018) 719–729. doi:10.1016/j.ijbiomac.2017.08.078.

SC
[111] M.C. Shivamadhu, K.S. Balaji, S. Jayarama, Haemostatic property of new Cystein protease(s)
from Sesbania grandiflora: It’s action on fibrinogens, Biocatal. Agric. Biotechnol. 12 (2017)
NU
10–14. doi:10.1016/j.bcab.2017.08.008.
[112] Y. Hua, S. Nair, Proteases in cardiometabolic diseases: Pathophysiology, molecular
mechanisms and clinical applications, Biochim. Biophys. Acta - Mol. Basis Dis. 1852 (2015)
MA

195–208. doi:10.1016/j.bbadis.2014.04.032.
[113] S. Li, X. Yang, S. Yang, M. Zhu, X. Wang, Technology prospecting on enzymes: application,
marketing and engineering., Comput. Struct. Biotechnol. J. 2 (2012) e201209017.
D

doi:10.5936/csbj.201209017.
E

[114] J. Ramundo, M. Gray, Enzymatic Wound Debridement, J. Wound, Ostomy Cont. Nurs. 35
PT

(2008) 273–280. doi:10.1097/01.WON.0000319125.21854.78.


[115] J.Q. Del Rosso, Application of protease technology in dermatology: rationale for incorporation
into skin care with initial observations on formulations designed for skin cleansing,
CE

maintenance of hydration, and restoration of the epidermal permeability barrier., J. Clin.


Aesthet. Dermatol. 6 (2013) 14–22. http://www.ncbi.nlm.nih.gov/pubmed/23882305 (accessed
AC

December 21, 2018).


[116] Proteolytic enzyme formulations, (2012). https://patents.google.com/patent/US8632769
(accessed December 21, 2018).
[117] Protease-based enzyme formulation for toothpaste - Creative Enzymes, (n.d.).
https://www.creative-enzymes.com/product/proteasebased-enzyme-formulation-for-
toothpaste_137.html.
[118] Protease (Formula 4) - 90 Vegetarian Capsules, (n.d.).
https://www.pureformulas.com/protease-formula-4-90-capsules-by-ness-enzymes.html.
[119] C. Díaz Blanco, A. Trifonov, G. Georgiev, T. Tzanov, Journal of molecular catalysis. B,
Enzymatic., Elsevier, 1995. http://agris.fao.org/agris-
ACCEPTED MANUSCRIPT

search/search.do?recordID=US201600060357 (accessed December 21, 2018).

PT
RI
SC
NU
MA
E D
PT
CE
AC
ACCEPTED MANUSCRIPT

Figure legends

Figure 1: Graphical abstract


Figure 2: Brief outline of protease isolation and purification strategies
Figure 3: Stain-removal by protease embedded detergent molecule
Figure 4: Influential ingredients in detergent making
Figure 5: The leather tanning process
Figure 6: Steps in cheese making

PT
Figure 7: Overall therapeutic application of proteases
Figure 8: Statistical illustration of historical development of research on protease

RI
Figure 9: Statistical illustration of historical development of industrial applications of protease

SC
NU
MA
E D
PT
CE
AC
ACCEPTED MANUSCRIPT

A systematic reconsideration on proteases

Poorani Gurumallesh, Kamalini Alagu, Baskar Ramakrishnan*, Shanmugaprakash Muthusamy

Figure 1: Graphical abstract

PT
RI
SC
NU
MA
E D
PT
CE
AC
ACCEPTED MANUSCRIPT

A systematic reconsideration on proteases

Poorani Gurumallesh, Kamalini Alagu, Baskar Ramakrishnan*, Shanmugaprakash Muthusamy

Figure 2: Brief outline of protease isolation and purification strategies

PT
RI
SC
NU
MA
E D
PT
CE
AC
ACCEPTED MANUSCRIPT

A systematic reconsideration on proteases

Poorani Gurumallesh, Kamalini Alagu, Baskar Ramakrishnan*, Shanmugaprakash Muthusamy

Figure 3: Stain-removal by protease embedded detergent molecule

PT
RI
SC
NU
MA
E D
PT
CE
AC
ACCEPTED MANUSCRIPT

A systematic reconsideration on proteases

Poorani Gurumallesh, Kamalini Alagu, Baskar Ramakrishnan*, Shanmugaprakash Muthusamy

Figure 4: Influential ingredients in detergent making

PT
RI
SC
NU
MA
E D
PT
CE
AC
ACCEPTED MANUSCRIPT

A systematic reconsideration on proteases

Poorani Gurumallesh, Kamalini Alagu, Baskar Ramakrishnan*, Shanmugaprakash Muthusamy

Figure 5: The leather tanning process

PT
RI
SC
NU
MA
E D
PT
CE
AC
ACCEPTED MANUSCRIPT

A systematic reconsideration on proteases

Poorani Gurumallesh, Kamalini Alagu, Baskar Ramakrishnan*, Shanmugaprakash Muthusamy

Figure 6: Steps in cheese making

PT
RI
SC
NU
MA
E D
PT
CE
AC
ACCEPTED MANUSCRIPT

A systematic reconsideration on proteases

Poorani Gurumallesh, Kamalini Alagu, Baskar Ramakrishnan*, Shanmugaprakash Muthusamy

Figure 7: Overall therapeutic application of proteases

PT
RI
SC
NU
MA
E D
PT
CE
AC
ACCEPTED MANUSCRIPT

A systematic reconsideration on proteases

Poorani Gurumallesh, Kamalini Alagu, Baskar Ramakrishnan*, Shanmugaprakash Muthusamy

Figure 8: Statistical illustration of historical development of research on protease

PT
Publications on extraction of protease from various sources
Source: SCOPUS database
40000

RI
35000
6248 5923 5860 5676
30000 6641

SC
6238
6047
25000 5489 6182 6049 6546 6571
5479 5549 5994
5362 5051 3973
20000 4376
3939
NU
3465 5029
15000 14707 14535 14594 14510
14278 14374
12708 13238
1000011648 12266 8568
MA

5000
5521 6264 6504 6791 6905 7250 7461 5781
4040 4559 4950
0
2008 2009 2010 2011 2012 2013 2014 2015 2016 2017 2018
D

Plant Animal Microbial rDNA


E
PT
CE
AC
ACCEPTED MANUSCRIPT

A systematic reconsideration on proteases

Poorani Gurumallesh, Kamalini Alagu, Baskar Ramakrishnan*, Shanmugaprakash Muthusamy

Figure 9: Statistical illustration of historical development of industrial applications of protease

PT
RI
SC
NU
MA
E D
PT
CE
AC

You might also like